101
|
Abstract
The poor uptake and limited effectiveness of seasonal influenza vaccines mean that influenza continues to create a significant burden of disease. It has been hypothesized that sex differences are present in responses to seasonal influenza vaccines, and that these differences may contribute to this poor vaccine success. This has led to the suggestion that vaccines should be tailored to an individual's biological sex. However, studies in this field are often low quality. Comprehensive analysis of the available literature reveals that there is insufficient evidence to support sex differences in vaccine immunogenicity, effectiveness, or efficacy. Nonetheless, differences in vaccine safety are consistently observed, with females reporting adverse events following immunization more frequently than males. Bias introduced by gender differences in passive reporting of adverse effects may underlie this phenomenon. Highly controlled studies are required in future before any conclusions can be made about potential sex differences in response to seasonal influenza vaccines.
Collapse
Affiliation(s)
- Lucy Denly
- Medical Sciences Division, University of Oxford, Oxford, UK
| |
Collapse
|
102
|
Quer G, Gadaleta M, Radin JM, Andersen KG, Baca-Motes K, Ramos E, Topol EJ, Steinhubl SR. The Physiologic Response to COVID-19 Vaccination. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021. [PMID: 33972954 DOI: 10.1101/2021.05.03.21256482] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Two mRNA vaccines and one adenovirus-based vaccine against SARS CoV-2 are currently being distributed at scale in the United States. Objective evidence of a specific individual's physiologic response to that vaccine are not routinely tracked but may offer insights into the acute immune response and personal and/or vaccine characteristics associated with that. We explored this possibility using a smartphone app-based research platform developed early in the pandemic that enabled volunteers (38,911 individuals between 25 March 2020 and 4 April 2021) to share their smartwatch and activity tracker data, as well as self-report, when appropriate, any symptoms, COVID-19 test results and vaccination dates and type. Of 4,110 individuals who reported at least one mRNA vaccination dose, 3,312 provided adequate resting heart rate data from the peri-vaccine period for analysis. We found changes in resting heart rate with respect to an individual baseline increased the days after vaccination, peaked on day 2, and returned to normal on day 6, with a much stronger effect after second dose with respect to first dose (average changes 1.6 versus 0.5 beats per minute). The changes were more pronounced for individuals who received the Moderna vaccine (on both doses), those who previously tested positive to COVID-19 (on dose 1), and for individuals aged <40 years, after adjusting for possible confounding factors. Taking advantage of continuous passive data from personal sensors could potentially enable the identification of a digital fingerprint of inflammation, which might prove useful as a surrogate for vaccine-induced immune response.
Collapse
|
103
|
Dhakal S, Loube J, Misplon JA, Lo CY, Creisher PS, Mulka KR, Deshpande S, Mitzner W, Klein SL, Epstein SL. Effect of an Adenovirus-Vectored Universal Influenza Virus Vaccine on Pulmonary Pathophysiology in a Mouse Model. J Virol 2021; 95:e02359-20. [PMID: 33627390 PMCID: PMC8104105 DOI: 10.1128/jvi.02359-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/17/2021] [Indexed: 11/20/2022] Open
Abstract
Current influenza vaccines, live attenuated or inactivated, do not protect against antigenically novel influenza A viruses (IAVs) of pandemic potential, which has driven interest in the development of universal influenza vaccines. Universal influenza vaccine candidates targeting highly conserved antigens of IAV nucleoprotein (NP) are promising as vaccines that induce T cell immunity, but concerns have been raised about the safety of inducing robust CD8 T cell responses in the lungs. Using a mouse model, we systematically evaluated effects of recombinant adenovirus vectors (rAd) expressing IAV NP (A/NP-rAd) or influenza B virus (IBV) NP (B/NP-rAd) on pulmonary inflammation and function after vaccination and following live IAV challenge. After A/NP-rAd or B/NP-rAd vaccination, female mice exhibited robust systemic and pulmonary vaccine-specific B cell and T cell responses and experienced no morbidity (e.g., body mass loss). Both in vivo pulmonary function testing and lung histopathology scoring revealed minimal adverse effects of intranasal rAd vaccination compared with unvaccinated mice. After IAV challenge, A/NP-rAd-vaccinated mice experienced significantly less morbidity, had lower pulmonary virus titers, and developed less pulmonary inflammation than unvaccinated or B/NP-rAd-vaccinated mice. Based on analysis of pulmonary physiology using detailed testing not previously applied to the question of T cell damage, mice protected by vaccination also had better lung function than controls. Results provide evidence that, in this model, adenoviral universal influenza vaccine does not damage pulmonary tissue. In addition, adaptive immunity, in particular, T cell immunity in the lungs, does not cause damage when restimulated but instead mitigates pulmonary damage following IAV infection.IMPORTANCE Respiratory viruses can emerge and spread rapidly before vaccines are available. It would be a tremendous advance to use vaccines that protect against whole categories of viruses, such as universal influenza vaccines, without the need to predict which virus will emerge. The nucleoprotein (NP) of influenza virus provides a target conserved among strains and is a dominant T cell target. In animals, vaccination to NP generates powerful T cell immunity and long-lasting protection against diverse influenza strains. Concerns have been raised, but not evaluated experimentally, that potent local T cell responses might damage the lungs. We analyzed lung function in detail in the setting of such a vaccination. Despite CD8 T cell responses in the lungs, lungs were not damaged and functioned normally after vaccination alone and were protected upon subsequent infection. This precedent provides important support for vaccines based on T cell-mediated protection, currently being considered for both influenza and SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Santosh Dhakal
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Jeffrey Loube
- Department of Environmental Health and Engineering, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Julia A Misplon
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Chia-Yun Lo
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Patrick S Creisher
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Kathleen R Mulka
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Sharvari Deshpande
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Wayne Mitzner
- Department of Environmental Health and Engineering, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Suzanne L Epstein
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
104
|
Ciarambino T, Barbagelata E, Corbi G, Ambrosino I, Politi C, Lavalle F, Ruggieri A, Moretti A. Gender differences in vaccine therapy: where are we in COVID-19 pandemic? Monaldi Arch Chest Dis 2021; 91. [PMID: 33840183 DOI: 10.4081/monaldi.2021.1669] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 03/24/2021] [Indexed: 11/23/2022] Open
Abstract
Vaccination is one of the greatest achievements of public health. Vaccination programs have contributed to the decline in mortality and morbidity of various infectious diseases. This review aims to investigate the impact of sex/gender on the vaccine acceptance, responses, and outcomes. The studies were identified by using PubMed, until 30th June 2020. The search was performed by using the following keywords: SARS-CoV-2, COVID-19, gender, sex, vaccine, adverse reaction. Clinical trials, retrospective and prospective studies were included. Studies written in languages other than English were excluded. Studies were included if gender differences in response to vaccination trials were reported. All selected studies were qualitatively analyzed. Innate recognition and response to viruses, as well as, adaptive immune responses during viral infections, differ between females and males. Unfortunately, a majority of vaccine trials have focused on healthy people, with ages between 18 to 65 years, excluding the elderly, pregnant women, post-menopausal female and children. In conclusion, it is apparent that the design of vaccines and vaccine strategies should be sex-specific, to reduce adverse reactions in females and increase immunogenicity in males. It should be mandatory to examine sex-related variables in pre-clinical and clinical vaccine trials, such as their crucial role for successful prevention of pandemic COVID-19.
Collapse
Affiliation(s)
| | | | - Graziamaria Corbi
- Department of Medicine and Health Sciences, University of Molise, Campobasso.
| | - Immacolata Ambrosino
- Local Healthcare Unit of Bari, Health District 10, Post-acute Care Triggiano, Bari.
| | - Cecilia Politi
- Head of Gender Medicine Area F.A.D.O.I., Internal Medicine Department, Isernia.
| | - Franco Lavalle
- Vicepresident OMCEO (Provincial order of surgeons and dentists), Bari.
| | - Anna Ruggieri
- Center for Gender Specific Medicine, Istituto Superiore Sanità, Rome.
| | | |
Collapse
|
105
|
Dhakal S, Ruiz-Bedoya CA, Zhou R, Creisher P, Villano J, Littlefield K, Castillo J, Marinho P, Jedlicka A, Ordonez A, Majewska N, Betenbaugh M, Flavahan K, Mueller A, Looney M, Quijada D, Mota F, Beck SE, Brockhurst JK, Braxton A, Castell N, D'Alessio F, Metcalf Pate KA, Karakousis PC, Mankowski JL, Pekosz A, Jain SK, Klein SL. Sex differences in lung imaging and SARS-CoV-2 antibody responses in a COVID-19 golden Syrian hamster model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.04.02.438292. [PMID: 33821269 PMCID: PMC8020969 DOI: 10.1101/2021.04.02.438292] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
In the ongoing coronavirus disease 2019 (COVID-19) pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), more severe outcomes are reported in males compared with females, including hospitalizations and deaths. Animal models can provide an opportunity to mechanistically interrogate causes of sex differences in the pathogenesis of SARS-CoV-2. Adult male and female golden Syrian hamsters (8-10 weeks of age) were inoculated intranasally with 10 5 TCID 50 of SARS-CoV-2/USA-WA1/2020 and euthanized at several time points during the acute (i.e., virus actively replicating) and recovery (i.e., after the infectious virus has been cleared) phases of infection. There was no mortality, but infected male hamsters experienced greater morbidity, losing a greater percentage of body mass, developing more extensive pneumonia as noted on chest computed tomography, and recovering more slowly than females. Treatment of male hamsters with estradiol did not alter pulmonary damage. Virus titers in respiratory tissues, including nasal turbinates, trachea, and lungs, and pulmonary cytokine concentrations, including IFNb and TNFa, were comparable between the sexes. However, during the recovery phase of infection, females mounted two-fold greater IgM, IgG, and IgA responses against the receptor-binding domain of the spike protein (S-RBD) in both plasma and respiratory tissues. Female hamsters also had significantly greater IgG antibodies against whole inactivated SARS-CoV-2 and mutant S-RBDs, as well as virus neutralizing antibodies in plasma. The development of an animal model to study COVID-19 sex differences will allow for a greater mechanistic understanding of the SARS-CoV-2 associated sex differences seen in the human population.
Collapse
|
106
|
Mauvais-Jarvis F, Berthold HK, Campesi I, Carrero JJ, Dakal S, Franconi F, Gouni-Berthold I, Heiman ML, Kautzky-Willer A, Klein SL, Murphy A, Regitz-Zagrosek V, Reue K, Rubin JB. Sex- and Gender-Based Pharmacological Response to Drugs. Pharmacol Rev 2021; 73:730-762. [PMID: 33653873 PMCID: PMC7938661 DOI: 10.1124/pharmrev.120.000206] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In humans, the combination of all sex-specific genetic, epigenetic, and hormonal influences of biologic sex produces different in vivo environments for male and female cells. We dissect how these influences of sex modify the pharmacokinetics and pharmacodynamics of multiple drugs and provide examples for common drugs acting on specific organ systems. We also discuss how gender of physicians and patients may influence the therapeutic response to drugs. We aim to highlight sex as a genetic modifier of the pharmacological response to drugs, which should be considered as a necessary step toward precision medicine that will benefit men and women. SIGNIFICANCE STATEMENT: This study discusses the influences of biologic sex on the pharmacokinetics and pharmacodynamics of drugs and provides examples for common drugs acting on specific organ systems. This study also discusses how gender of physicians and patients influence the therapeutic response to drugs.
Collapse
Affiliation(s)
- Franck Mauvais-Jarvis
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Heiner K Berthold
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Ilaria Campesi
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Juan-Jesus Carrero
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Santosh Dakal
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Flavia Franconi
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Ioanna Gouni-Berthold
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Mark L Heiman
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Alexandra Kautzky-Willer
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Sabra L Klein
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Anne Murphy
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Vera Regitz-Zagrosek
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Karen Reue
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Joshua B Rubin
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| |
Collapse
|
107
|
Matera MG, Ora J, Calzetta L, Rogliani P, Cazzola M. Sex differences in COPD management. Expert Rev Clin Pharmacol 2021; 14:323-332. [PMID: 33560876 DOI: 10.1080/17512433.2021.1888713] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Introduction: A growing body of evidence indicates that prevalence of chronic obstructive pulmonary disease (COPD) has been increasing more rapidly among women compared to men, but the available data on the differences between the sexes in the responses to the various COPD therapies are still scarce.Areas covered: The aim of this narrative review is to provide an overview of current knowledge on sex differences in COPD management.Expert opinion: There is no solid evidence of sex differences in response to usual COPD treatments but there are sex-related differences in management of patients with a clinical diagnosis of COPD. It is difficult to explain the reason for these differences, but most likely they are due to local prescribing habits, rather than solid scientific reasons. However, there are also signals of different sex-related responses, the qualification and quantification of which is difficult with the information currently available. These signals should lead to the inclusion of more women in clinical trials, but also to the design of prospective clinical studies to assess the possible differences linked to sex in COPD treatment responses, whose identification is an important step toward the definition of personalized COPD therapy.
Collapse
Affiliation(s)
- Maria Gabriella Matera
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Josuel Ora
- Respiratory Diseases Unit, "Tor Vergata" University Hospital, Rome, Italy
| | - Luigino Calzetta
- Unit of Respiratory Disease and Lung Function Dept. Medicine and Surgery, University of Parma, ParmaItaly
| | - Paola Rogliani
- Respiratory Diseases Unit, "Tor Vergata" University Hospital, Rome, Italy.,Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Mario Cazzola
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
108
|
Ursin RL, Liu H, Powell HR, Westerbeck JW, Shaw-Saliba K, Sylvia KE, Fenstermacher KJ, Mehoke T, Thielen P, Rothman RE, Pekosz A, Klein SL. Differential Antibody Recognition of H3N2 Vaccine and Seasonal Influenza Virus Strains Based on Age, Vaccine Status, and Sex in the 2017-2018 Season. J Infect Dis 2021; 222:1371-1382. [PMID: 32496543 DOI: 10.1093/infdis/jiaa289] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 05/22/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND An antigenic mismatch between the vaccine and circulating H3N2 strains was hypothesized to contribute to the severity of the 2017-2018 season in North America. METHODS Serum and nasal washes were collected from influenza positive and negative patients during the 2017-2018 season to determine neutralizing antibody (nAb) titers and for influenza virus sequencing, respectively. RESULTS The circulating and vaccine H3N2 virus strains were different clades, with the vaccine strain being clade 3C.2a and the circulating viruses being 3C.2a2 or 3C.3a. At enrollment, both the H3N2 negative and positive patients had greater nAb titers to the egg-adapted vaccine virus compared to the cell-grown vaccine but the H3N2-negative population had significantly greater titers to the circulating 3C.2a2. Among H3N2-positive patients, vaccination, younger age, and female sex were associated with greater nAb responses to the egg-adapted vaccine H3N2 virus but not to the cell-grown vaccine or circulating viruses. CONCLUSIONS For the 2017-2018 circulating viruses, mutations introduced by egg adaptation decreased vaccine efficacy. No increased protection was afforded by vaccination, younger age, or female sex against 2017-2018 circulating H3N2 viruses.
Collapse
Affiliation(s)
- Rebecca L Ursin
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Hsuan Liu
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Harrison R Powell
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Jason W Westerbeck
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Kathryn Shaw-Saliba
- Department of Emergency Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kristyn E Sylvia
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Katherine J Fenstermacher
- Department of Emergency Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tom Mehoke
- Johns Hopkins Applied Physics Laboratory, Laurel, Maryland, USA
| | - Peter Thielen
- Johns Hopkins Applied Physics Laboratory, Laurel, Maryland, USA
| | - Richard E Rothman
- Department of Emergency Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Sabra L Klein
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.,W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
109
|
Murdock BJ, Goutman SA, Boss J, Kim S, Feldman EL. Amyotrophic Lateral Sclerosis Survival Associates With Neutrophils in a Sex-specific Manner. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/2/e953. [PMID: 33531377 PMCID: PMC8057067 DOI: 10.1212/nxi.0000000000000953] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 11/13/2020] [Indexed: 12/11/2022]
Abstract
Objective To determine whether neutrophils contribute to amyotrophic lateral sclerosis
(ALS) progression, we tested the association of baseline neutrophil count on
ALS survival, whether the effect was sex specific, and whether neutrophils
accumulate in the spinal cord. Methods A prospective cohort study was conducted between June 22, 2011, and October
30, 2019. Blood leukocytes were isolated from ALS participants and
neutrophil levels assessed by flow cytometry. Participant survival outcomes
were analyzed by groups (<2 × 106, 2–4 ×
106, and >4 × 106 neutrophils/mL) with
adjustments for relevant ALS covariates and by sex. Neutrophil levels were
assessed from CNS tissue from a subset of participants. Results A total of 269 participants with ALS within 2 years of an ALS diagnosis were
included. Participants with baseline neutrophil counts over 4 ×
106/mL had a 2.1 times higher mortality rate than those with
a neutrophil count lower than 2 × 106/mL (95% CI:
1.3–3.5, p = 0.004) when adjusting for age,
sex, and other covariates. This effect was more pronounced in females, with
a hazard ratio of 3.8 (95% CI: 1.8–8.2, p =
0.001) in the >4 × 106/mL vs <2 ×
106/mL group. Furthermore, ALS participants (n = 8) had
increased neutrophils in cervical (p = 0.049) and
thoracic (p = 0.022) spinal cord segments compared
with control participants (n = 8). Conclusions Higher neutrophil counts early in ALS associate with a shorter survival in
female participants. Furthermore, neutrophils accumulate in ALS spinal cord
supporting a pathophysiologic correlate. These data justify the
consideration of immunity and sex for personalized therapeutic development
in ALS. Classification of Evidence This study provides Class III evidence that in female participants with ALS,
higher baseline neutrophil counts are associated with shorter survival.
Collapse
Affiliation(s)
- Benjamin J Murdock
- From the Department of Neurology (B.J.M., S.A.G., E.L.F.), and Department of Biostatistics (J.B., S.K.), School of Public Health, University of Michigan, Ann Arbor
| | - Stephen A Goutman
- From the Department of Neurology (B.J.M., S.A.G., E.L.F.), and Department of Biostatistics (J.B., S.K.), School of Public Health, University of Michigan, Ann Arbor.
| | - Jonathan Boss
- From the Department of Neurology (B.J.M., S.A.G., E.L.F.), and Department of Biostatistics (J.B., S.K.), School of Public Health, University of Michigan, Ann Arbor
| | - Sehee Kim
- From the Department of Neurology (B.J.M., S.A.G., E.L.F.), and Department of Biostatistics (J.B., S.K.), School of Public Health, University of Michigan, Ann Arbor
| | - Eva L Feldman
- From the Department of Neurology (B.J.M., S.A.G., E.L.F.), and Department of Biostatistics (J.B., S.K.), School of Public Health, University of Michigan, Ann Arbor
| |
Collapse
|
110
|
Rebuli ME, Glista-Baker E, Hoffman JR, Duffney PF, Robinette C, Speen AM, Pawlak EA, Dhingra R, Noah TL, Jaspers I. Electronic-Cigarette Use Alters Nasal Mucosal Immune Response to Live-attenuated Influenza Virus. A Clinical Trial. Am J Respir Cell Mol Biol 2021; 64:126-137. [PMID: 33095645 PMCID: PMC7781000 DOI: 10.1165/rcmb.2020-0164oc] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Inhalation of tobacco smoke has been linked to increased risk of viral infection, such as influenza. Inhalation of electronic-cigarette (e-cigarette) aerosol has also recently been linked to immune suppression within the respiratory tract, specifically the nasal mucosa. We propose that changes in the nasal mucosal immune response modify antiviral host-defense responses in e-cigarette users. Nonsmokers, cigarette smokers, and e-cigarette users were inoculated with live-attenuated influenza virus (LAIV) to safely examine the innate immune response to influenza infection. Before and after LAIV inoculation, we collected nasal epithelial-lining fluid, nasal lavage fluid, nasal-scrape biopsy specimens, urine, and blood. Endpoints examined include cytokines and chemokines, influenza-specific IgA, immune-gene expression, and markers of viral load. Statistical analysis included primary comparisons of cigarette and e-cigarette groups with nonsmokers, as well as secondary analysis of demographic factors as potential modifiers. Markers of viral load did not differ among the three groups. Nasal-lavage-fluid anti-LAIV IgA levels increased in nonsmokers after LAIV inoculation but did not increase in e-cigarette users and cigarette smokers. LAIV-induced gene-expression changes in nasal biopsy specimens differed in cigarette smokers and e-cigarette users as compared with nonsmokers, with a greater number of genes changed in e-cigarette users, mostly resulting in decreased expression. The top downregulated genes in cigarette smokers were SMPD3, NOS2A, and KLRB1, and the top downregulated genes in e-cigarette users were MR1, NT5E, and HRAS. Similarly, LAIV-induced cytokine levels in nasal epithelial-lining fluid differed among the three groups, including decreased antiviral host-defense mediators (IFNγ, IL6, and IL12p40). We also detected that sex interacted with tobacco-product exposure to modify LAIV-induced immune-gene expression. Our results demonstrate that e-cigarette use altered nasal LAIV-induced immune responses, including gene expression, cytokine and chemokine release, and LAIV-specific IgA levels. Together, these data suggest that e-cigarette use induces changes in the nasal mucosa that are consistent with the potential for altered respiratory antiviral host-defense function. Clinical trial registered with www.clinicaltrials.gov (NCT 02019745).
Collapse
Affiliation(s)
- Meghan E Rebuli
- Curriculum in Toxicology and Environmental Medicine.,Center for Environmental Medicine, Asthma and Lung Biology, and.,Department of Pediatrics, School of Medicine
| | | | - Jessica R Hoffman
- Curriculum for the Environment and Ecology, College of Arts and Sciences
| | | | | | - Adam M Speen
- Curriculum in Toxicology and Environmental Medicine
| | - Erica A Pawlak
- Center for Environmental Medicine, Asthma and Lung Biology, and
| | - Radhika Dhingra
- Institute for Environmental Health Solutions, and.,Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Terry L Noah
- Center for Environmental Medicine, Asthma and Lung Biology, and.,Department of Pediatrics, School of Medicine
| | - Ilona Jaspers
- Curriculum in Toxicology and Environmental Medicine.,Center for Environmental Medicine, Asthma and Lung Biology, and.,Department of Pediatrics, School of Medicine.,Institute for Environmental Health Solutions, and
| |
Collapse
|
111
|
Ciarambino T, Para O, Giordano M. Immune system and COVID-19 by sex differences and age. WOMEN'S HEALTH (LONDON, ENGLAND) 2021; 17:17455065211022262. [PMID: 34096383 PMCID: PMC8188967 DOI: 10.1177/17455065211022262] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/19/2021] [Accepted: 05/06/2021] [Indexed: 12/12/2022]
Abstract
In COVID-19 disease, are reported gender differences in relation to severity and death. The aim of this review is to highlight gender differences in the immune response to COVID-19. The included studies were identified using PubMed, until 30 October 2020. The search included the following keywords: SARS-CoV-2, COVID-19, gender, age, sex, and immune system. Literature described that females compared to males have greater inflammatory, antiviral, and humoral immune responses. In female, estrogen is a potential ally to alleviate SARS-COV-2 disease. In male, testosterone reduces vaccination response and depresses the cytokine response. In the older patients, and in particular, in female older patients, it has been reported a progressive functional decline in the immune systems. Differences by gender were reported in infection diseases, including SARS-CoV-2. These data should be confirmed by the other epidemiological studies.
Collapse
Affiliation(s)
- Tiziana Ciarambino
- Department of Internal Medicine, Marcianise Hospital, ASL Caserta, Italy
| | - Ombretta Para
- Department of Internal Medicine, Careggi Hospital, University of Florence, Florence, Italy
| | - Mauro Giordano
- Department of Internal Medicine, University of Campania, L. Vanvitelli, Naples, Italy
| |
Collapse
|
112
|
Klingler J, Weiss S, Itri V, Liu X, Oguntuyo KY, Stevens C, Ikegame S, Hung CT, Enyindah-Asonye G, Amanat F, Baine I, Arinsburg S, Bandres JC, Kojic EM, Stoever J, Jurczyszak D, Bermudez-Gonzalez M, Nádas A, Liu S, Lee B, Zolla-Pazner S, Hioe CE. Role of Immunoglobulin M and A Antibodies in the Neutralization of Severe Acute Respiratory Syndrome Coronavirus 2. J Infect Dis 2020; 223:957-970. [PMID: 33367897 PMCID: PMC7798948 DOI: 10.1093/infdis/jiaa784] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 12/18/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has infected millions of people globally. Virus infection requires the receptor-binding domain (RBD) of the spike protein. Although studies have demonstrated anti-spike and -RBD antibodies to be protective in animal models, and convalescent plasma as a promising therapeutic option, little is known about immunoglobulin isotypes capable of blocking infection. METHODS We studied spike- and RBD-specific immunoglobulin isotypes in convalescent and acute plasma/serum samples using a multiplex bead assay. We also determined virus neutralization activities in plasma and serum samples, and purified immunoglobulin fractions using a vesicular stomatitis pseudovirus assay. RESULTS Spike- and RBD-specific immunoglobulin (Ig) M, IgG1, and IgA1 were produced by all or nearly all subjects at variable levels and detected early after infection. All samples displayed neutralizing activity. Regression analyses revealed that IgM and IgG1 contributed most to neutralization, consistent with IgM and IgG fractions' neutralization potency. IgA also exhibited neutralizing activity, but with lower potency. CONCLUSION IgG, IgM, and IgA are critical components of convalescent plasma used for treatment of coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Jéromine Klingler
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA,James J. Peters VA Medical Center, Bronx, New York, USA
| | - Svenja Weiss
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA,James J. Peters VA Medical Center, Bronx, New York, USA
| | - Vincenza Itri
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Xiaomei Liu
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA,James J. Peters VA Medical Center, Bronx, New York, USA
| | - Kasopefoluwa Y Oguntuyo
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Christian Stevens
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Satoshi Ikegame
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Chuan-Tien Hung
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Gospel Enyindah-Asonye
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Fatima Amanat
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ian Baine
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Suzanne Arinsburg
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Erna Milunka Kojic
- Division of Infectious Diseases, Department of Medicine, Mount Sinai West and Morningside, New York, USA
| | - Jonathan Stoever
- Pulmonary and Critical Care Medicine, Mount Sinai West, New York, USA
| | - Denise Jurczyszak
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Maria Bermudez-Gonzalez
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Arthur Nádas
- Department of Environment Medicine, NYU School of Medicine, New York, New York, USA
| | - Sean Liu
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA,Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Benhur Lee
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Susan Zolla-Pazner
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA,Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Catarina E Hioe
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA,James J. Peters VA Medical Center, Bronx, New York, USA,Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA,Correspondence: Catarina E. Hioe, () ()
| |
Collapse
|
113
|
Chudnovets A, Liu J, Narasimhan H, Liu Y, Burd I. Role of Inflammation in Virus Pathogenesis during Pregnancy. J Virol 2020; 95:e01381-19. [PMID: 33115865 PMCID: PMC7944452 DOI: 10.1128/jvi.01381-19] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Viral infections during pregnancy lead to a spectrum of maternal and fetal outcomes, ranging from asymptomatic disease to more critical conditions presenting with severe maternal morbidity, stillbirth, preterm birth, intrauterine growth restriction, and fetal congenital anomalies, either apparent at birth or later in life. In this article, we review the pathogenesis of several viral infections that are particularly relevant in the context of pregnancy and intrauterine inflammation. Understanding the diverse mechanisms employed by viral pathogens as well as the repertoire of immune responses induced in the mother may help to establish novel therapeutic options to attenuate changes in the maternal-fetal interface and prevent adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Anna Chudnovets
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jin Liu
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Harish Narasimhan
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Yang Liu
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Irina Burd
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
114
|
Klingler J, Weiss S, Itri V, Liu X, Oguntuyo KY, Stevens C, Ikegame S, Hung CT, Enyindah-Asonye G, Amanat F, Baine I, Arinsburg S, Bandres JC, Kojic EM, Stoever J, Jurczyszak D, Bermudez-Gonzalez M, Nádas A, Liu S, Lee B, Zolla-Pazner S, Hioe CE. Role of IgM and IgA Antibodies in the Neutralization of SARS-CoV-2. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.08.18.20177303. [PMID: 33173891 PMCID: PMC7654883 DOI: 10.1101/2020.08.18.20177303] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND SARS-CoV-2 has infected millions of people globally. Virus infection requires the receptor-binding domain (RBD) of the spike protein. Although studies have demonstrated anti-spike and - RBD antibodies to be protective in animal models, and convalescent plasma as a promising therapeutic option, little is known about immunoglobulin (Ig) isotypes capable of blocking infection. METHODS We studied spike- and RBD-specific Ig isotypes in convalescent and acute plasma/sera using a multiplex bead assay. We also determined virus neutralization activities in plasma, sera, and purified Ig fractions using a VSV pseudovirus assay. RESULTS Spike- and RBD-specific IgM, IgG1, and IgA1 were produced by all or nearly all subjects at variable levels and detected early after infection. All samples displayed neutralizing activity. Regression analyses revealed that IgM and IgG1 contributed most to neutralization, consistent with IgM and IgG fractions' neutralization potency. IgA also exhibited neutralizing activity, but with lower potency. CONCLUSION IgG, IgM and IgA are critical components of convalescent plasma used for COVID-19 treatment.
Collapse
Affiliation(s)
- Jéromine Klingler
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY USA
- James J. Peters VA Medical Center, Bronx, NY, USA
| | - Svenja Weiss
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY USA
- James J. Peters VA Medical Center, Bronx, NY, USA
| | - Vincenza Itri
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Xiaomei Liu
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY USA
- James J. Peters VA Medical Center, Bronx, NY, USA
| | | | - Christian Stevens
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Satoshi Ikegame
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chuan-Tien Hung
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gospel Enyindah-Asonye
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Fatima Amanat
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ian Baine
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Suzanne Arinsburg
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Erna Milunka Kojic
- Division of Infectious Diseases, Department of Medicine, Mount Sinai West and Morningside, NY, USA
| | | | - Denise Jurczyszak
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Arthur Nádas
- Department of Environment Medicine, NYU School of Medicine, New York, NY, USA
| | - Sean Liu
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benhur Lee
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Susan Zolla-Pazner
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Catarina E. Hioe
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY USA
- James J. Peters VA Medical Center, Bronx, NY, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
115
|
Honce R, Wohlgemuth N, Meliopoulos VA, Short KR, Schultz-Cherry S. Influenza in High-Risk Hosts-Lessons Learned from Animal Models. Cold Spring Harb Perspect Med 2020; 10:a038604. [PMID: 31871227 PMCID: PMC7706577 DOI: 10.1101/cshperspect.a038604] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Factoring significantly into the global burden of influenza disease are high-risk populations that suffer the bulk of infections. Classically, the very young, very old, and pregnant women have been identified as high-risk populations; however, recent research has uncovered several other conditions that contribute to severe infection. By using varied animal models, researchers have identified molecular mechanisms underpinning the increased likelihood for infection due to obesity and malnourishment, as well as insight into the role sex hormones play in antiviral immunity in males, in females, and across the life span. Additionally, novel comorbidity models have helped elucidate the role of chronic infectious and genetic diseases in influenza virus pathogenesis. Animal models play a vital role in understanding the contribution of host factors to influenza severity and immunity. An in-depth understanding of these host factors represents an important step in reducing the burden of influenza among the growing number of people living with one or more chronic medical conditions.
Collapse
Affiliation(s)
- Rebekah Honce
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee 38105-3678, USA
- Integrated Program in Biomedical Sciences, Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - Nicholas Wohlgemuth
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee 38105-3678, USA
| | - Victoria A Meliopoulos
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee 38105-3678, USA
| | - Kirsty R Short
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Stacey Schultz-Cherry
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee 38105-3678, USA
| |
Collapse
|
116
|
Acheampong DO, Barffour IK, Boye A, Aninagyei E, Ocansey S, Morna MT. Male predisposition to severe COVID-19: Review of evidence and potential therapeutic prospects. Biomed Pharmacother 2020; 131:110748. [PMID: 33152916 PMCID: PMC7480230 DOI: 10.1016/j.biopha.2020.110748] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/04/2020] [Accepted: 09/07/2020] [Indexed: 02/07/2023] Open
Abstract
The severe form of COVID-19 has significant sex disparities, with high fatalities commonly reported among males than females. The incidence of COVID-19 has also been higher in males compared with their female counterparts. This trend could be attributed to a better responsive and robust immune system in females. Cytokine storm is one of the pathophysiological features of severe COVID-19, and it occurs as a result of over-activation of immune cells leading to severe inflammation and tissue damage. Nevertheless, it is well modulated in females compared to their male counterparts. Severe inflammation in males is reported to facilitate progression of mild to severe COVID-19. The sex hormones, estrogens and androgens which exist in varying functional levels respectively in females and males are cited as the underlying cause for the differential immune response to COVID-19. Evidence abounds that estrogen modulate the immune system to protect females from severe inflammation and for that matter severe COVID-19. On the contrary, androgen has been implicated in over-activation of immune cells, cytokine storm and the attendant severe inflammation, which perhaps predispose males to severe COVID-19. In this review efforts are made to expand understanding and explain the possible roles of the immune system, the sex hormones and the angiotensin-converting enzyme (ACE) systems in male bias to severe COVID-19. Also, this review explores possible therapeutic avenues including androgen deprivation therapy (ADT), estrogen-based therapy, and ACE inhibitors for consideration in the fight against COVID-19.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Androgen Antagonists/pharmacology
- Androgen Antagonists/therapeutic use
- Angiotensin-Converting Enzyme 2
- Angiotensin-Converting Enzyme Inhibitors/pharmacology
- Angiotensin-Converting Enzyme Inhibitors/therapeutic use
- Animals
- Betacoronavirus/physiology
- COVID-19
- Child
- Child, Preschool
- Coronavirus Infections/complications
- Coronavirus Infections/drug therapy
- Coronavirus Infections/epidemiology
- Coronavirus Infections/immunology
- Coronavirus Infections/therapy
- Disease Susceptibility
- Female
- Gonadal Steroid Hormones/physiology
- Humans
- Immunity, Innate
- Infant
- Infant, Newborn
- Inflammation
- Male
- Mice
- Middle Aged
- Pandemics
- Peptidyl-Dipeptidase A/physiology
- Pneumonia, Viral/complications
- Pneumonia, Viral/epidemiology
- Pneumonia, Viral/immunology
- Pneumonia, Viral/therapy
- Prostatic Neoplasms/complications
- Prostatic Neoplasms/drug therapy
- Protein Disulfide-Isomerases/physiology
- Receptors, Cell Surface/physiology
- Receptors, Virus/physiology
- SARS-CoV-2
- Sex Distribution
- Smoking/adverse effects
- Young Adult
- COVID-19 Drug Treatment
Collapse
Affiliation(s)
- Desmond Omane Acheampong
- Department of Biomedical Sciences, School of Allied Health Sciences, College of Health and Allied Sciences, University of Cape Coast, Ghana.
| | - Isaac Kyei Barffour
- Department of Biomedical Sciences, School of Allied Health Sciences, College of Health and Allied Sciences, University of Cape Coast, Ghana
| | - Alex Boye
- Department of Medical Laboratory Science, School of Allied Health Sciences, College of Health and Allied Sciences, University of Cape Coast, Ghana
| | - Enoch Aninagyei
- Department of Biomedical Sciences, School of Basic and Biomedical Sciences, University of Allied Health Sciences, Ho, Ghana
| | - Stephen Ocansey
- Department of Optometry and Vision Science, School of Allied Health Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Martin Tangnaa Morna
- Department of Surgery, School of Medical Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| |
Collapse
|
117
|
Dingens AS, Crawford KHD, Adler A, Steele SL, Lacombe K, Eguia R, Amanat F, Walls AC, Wolf CR, Murphy M, Pettie D, Carter L, Qin X, King NP, Veesler D, Krammer F, Dickerson JA, Chu HY, Englund JA, Bloom JD. Serological identification of SARS-CoV-2 infections among children visiting a hospital during the initial Seattle outbreak. Nat Commun 2020; 11:4378. [PMID: 32873791 PMCID: PMC7463158 DOI: 10.1038/s41467-020-18178-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 08/07/2020] [Indexed: 11/09/2022] Open
Abstract
Children are strikingly underrepresented in COVID-19 case counts. In the United States, children represent 22% of the population but only 1.7% of confirmed SARS-CoV-2 cases as of April 2, 2020. One possibility is that symptom-based viral testing is less likely to identify infected children, since they often experience milder disease than adults. Here, to better assess the frequency of pediatric SARS-CoV-2 infection, we serologically screen 1,775 residual samples from Seattle Children's Hospital collected from 1,076 children seeking medical care during March and April of 2020. Only one child was seropositive in March, but seven were seropositive in April for a period seroprevalence of ≈1%. Most seropositive children (6/8) were not suspected of having had COVID-19. The sera of seropositive children have neutralizing activity, including one that neutralized at a dilution > 1:18,000. Therefore, an increasing number of children seeking medical care were infected by SARS-CoV-2 during the early Seattle outbreak despite few positive viral tests.
Collapse
Affiliation(s)
- Adam S Dingens
- Basic Sciences and Computational Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Katharine H D Crawford
- Basic Sciences and Computational Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA, 98195, USA
| | - Amanda Adler
- Division of Infectious Disease, Seattle Children's Hospital, Seattle, WA, 98105, USA
| | - Sarah L Steele
- Division of Infectious Disease, Seattle Children's Hospital, Seattle, WA, 98105, USA
| | - Kirsten Lacombe
- Division of Infectious Disease, Seattle Children's Hospital, Seattle, WA, 98105, USA
| | - Rachel Eguia
- Basic Sciences and Computational Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Fatima Amanat
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Alexandra C Walls
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
| | - Caitlin R Wolf
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, 98195, USA
| | - Michael Murphy
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Deleah Pettie
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Lauren Carter
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Xuan Qin
- Division of Infectious Disease, Seattle Children's Hospital, Seattle, WA, 98105, USA
| | - Neil P King
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jane A Dickerson
- Division of Infectious Disease, Seattle Children's Hospital, Seattle, WA, 98105, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Helen Y Chu
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, 98195, USA
| | - Janet A Englund
- Division of Infectious Disease, Seattle Children's Hospital, Seattle, WA, 98105, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, 98195, USA.
| | - Jesse D Bloom
- Basic Sciences and Computational Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA.
- Howard Hughes Medical Institute, Seattle, WA, 98103, USA.
| |
Collapse
|
118
|
Mauvais-Jarvis F, Bairey Merz N, Barnes PJ, Brinton RD, Carrero JJ, DeMeo DL, De Vries GJ, Epperson CN, Govindan R, Klein SL, Lonardo A, Maki PM, McCullough LD, Regitz-Zagrosek V, Regensteiner JG, Rubin JB, Sandberg K, Suzuki A. Sex and gender: modifiers of health, disease, and medicine. Lancet 2020; 396:565-582. [PMID: 32828189 PMCID: PMC7440877 DOI: 10.1016/s0140-6736(20)31561-0] [Citation(s) in RCA: 1240] [Impact Index Per Article: 248.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 06/12/2020] [Accepted: 06/16/2020] [Indexed: 02/09/2023]
Abstract
Clinicians can encounter sex and gender disparities in diagnostic and therapeutic responses. These disparities are noted in epidemiology, pathophysiology, clinical manifestations, disease progression, and response to treatment. This Review discusses the fundamental influences of sex and gender as modifiers of the major causes of death and morbidity. We articulate how the genetic, epigenetic, and hormonal influences of biological sex influence physiology and disease, and how the social constructs of gender affect the behaviour of the community, clinicians, and patients in the health-care system and interact with pathobiology. We aim to guide clinicians and researchers to consider sex and gender in their approach to diagnosis, prevention, and treatment of diseases as a necessary and fundamental step towards precision medicine, which will benefit men's and women's health.
Collapse
Affiliation(s)
- Franck Mauvais-Jarvis
- Diabetes Discovery & Sex-Based Medicine Laboratory, Section of Endocrinology, John W Deming Department of Medicine, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, LA, USA.
| | - Noel Bairey Merz
- Barbra Streisand Women's Heart Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA, USA
| | - Peter J Barnes
- National Heart & Lung Institute, Imperial College London, London, UK
| | - Roberta D Brinton
- Department of Pharmacology and Department of Neurology, College of Medicine, Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, USA
| | - Juan-Jesus Carrero
- Department of Medical Epidemiology and Biostatistics and Center for Gender Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Dawn L DeMeo
- Channing Division of Network Medicine and the Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Geert J De Vries
- Neuroscience Institute and Department of Biology, Georgia State University, Atlanta, GA, USA
| | - C Neill Epperson
- Department of Psychiatry, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Ramaswamy Govindan
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Sabra L Klein
- W Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Amedeo Lonardo
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, Ospedale Civile di Baggiovara, Modena, Italy
| | - Pauline M Maki
- Department of Psychiatry, Department of Psychology, and Department of Obstetrics & Gynecology, University of Illinois at Chicago, Chicago, IL, USA
| | - Louise D McCullough
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Vera Regitz-Zagrosek
- Berlin Institute of Gender Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany; Department of Cardiology, University Hospital Zürich, University of Zürich, Switzerland
| | - Judith G Regensteiner
- Center for Women's Health Research, Divisions of General Internal Medicine and Cardiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Joshua B Rubin
- Department of Medicine, Department of Paediatrics, and Department of Neuroscience, Washington University School of Medicine St Louis, MO, USA
| | - Kathryn Sandberg
- Center for the Study of Sex Differences in Health, Aging and Disease, Georgetown University, Washington, DC, USA
| | - Ayako Suzuki
- Division of Gastroenterology, Duke University Medical Center Durham, NC, USA; Durham VA Medical Center, Durham, NC, USA
| |
Collapse
|
119
|
Papadopoulos V, Li L, Samplaski M. Why does COVID-19 kill more elderly men than women? Is there a role for testosterone? Andrology 2020; 9:65-72. [PMID: 32681716 PMCID: PMC7404939 DOI: 10.1111/andr.12868] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/08/2020] [Accepted: 07/13/2020] [Indexed: 01/08/2023]
Abstract
Background Recent epidemiological data indicate that there may be a gender predisposition to COVID‐19, with men predisposed to being most severely affected, and older men accounting for most deaths. Objectives Provide a review of the research literature, propose hypotheses, and therapies based on the potential link between testosterone (T) and COVID‐19 induced mortality in elderly men. Materials and Methods A search of publications in academic electronic databases, and government and public health organization web sites on T, aging, inflammation, severe acute respiratory syndrome (SARS) due to coronavirus (CoV) 2 (SARS‐CoV‐2) infection, and COVID‐19 disease state and outcomes was performed. Results The link between T, the immune system, and male aging is well‐established, as is the progressive decline in T levels with aging. In women, T levels drop before menopause and variably increase with advanced age. Elevated IL‐6 is a characteristic biomarker of patients infected with COVID‐19 and has been linked to the development of the acute respiratory distress syndrome (ARDS). Thus far, half of the admitted COVID‐19 patients developed ARDS, half of these patients died, and elderly male patients have been more likely to develop ARDS and die. Low T is associated with ARDS. These data suggest that low T levels may exacerbate the severity of COVID‐19 infection in elderly men. It may also stand to reason that normal T levels may offer some protection against COVID‐19. SARS‐CoV‐2 binds to the angiotensin‐converting enzyme 2, present in high levels in the testis. Conclusion At present, it is not known whether low T levels in aging hypogonadal males create a permissive environment for severe responses to COVID‐19 infection or if the virus inhibits androgen formation. Given the preponderance of COVID‐19 related mortality in elderly males, additional testing for gonadal function and treatment with T may be merited.
Collapse
Affiliation(s)
- Vassilios Papadopoulos
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Lu Li
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Mary Samplaski
- Keck School of Medicine, Institute of Urology, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
120
|
Abstract
A male bias in mortality has emerged in the COVID-19 pandemic, which is consistent with the pathogenesis of other viral infections. Biological sex differences may manifest themselves in susceptibility to infection, early pathogenesis, innate viral control, adaptive immune responses or the balance of inflammation and tissue repair in the resolution of infection. We discuss available sex-disaggregated epidemiological data from the COVID-19 pandemic, introduce sex-differential features of immunity and highlight potential sex differences underlying COVID-19 severity. We propose that sex differences in immunopathogenesis will inform mechanisms of COVID-19, identify points for therapeutic intervention and improve vaccine design and increase vaccine efficacy. Why are males more susceptible to severe COVID-19 than females? In this Perspective, Sabra Klein and colleagues consider the sex differences in the immune system that may contribute to this sex bias.
Collapse
|
121
|
Aaby P, Benn CS, Flanagan KL, Klein SL, Kollmann TR, Lynn DJ, Shann F. The non-specific and sex-differential effects of vaccines. Nat Rev Immunol 2020; 20:464-470. [PMID: 32461674 PMCID: PMC7252419 DOI: 10.1038/s41577-020-0338-x] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2020] [Indexed: 12/27/2022]
Abstract
The textbook view of vaccination is that it functions to induce immune memory of the specific pathogen components of the vaccine, leading to a quantitatively and qualitatively better response if the host is exposed to infection with the same pathogen. However, evidence accumulated over the past few decades increasingly suggests that vaccines can also have non-specific effects on unrelated infections and diseases, with important implications for childhood mortality particularly in low-income settings. Furthermore, many of these non-specific effects, as well as the pathogen-specific effects, of vaccines show differences between the sexes. Here, members of the Optimmunize consortium discuss the evidence for and potential mechanisms of non-specific and sex-differential effects of vaccines, as well as their potential policy implications. Given that the non-specific effects of some vaccines are now being tested for their ability to protect against COVID-19, the authors also comment on the broader implications of these trials. In this Viewpoint article, members of the Optimmunize consortium discuss the evidence for non-specific and sex-differential effects of vaccines and how this information might inform vaccine design and policy, including in relation to the COVID-19 pandemic. Peter Aaby was trained as an anthropologist but has built a large health surveillance system in Guinea-Bissau since 1978, focusing on the high levels of child mortality there. Crowding and intensive exposure to measles were key determinants of child mortality. This led to vaccine research and the discovery of the non-specific effects of measles vaccine. Christine Stabell Benn is a professor in global health at the University of Southern Denmark. She conducts epidemiological and immunological studies of vaccines and vitamin A, with a focus on their real-life effects on overall health in Africa and Denmark. She formulated the hypothesis that these health interventions with immunomodulatory effects interact, often in a sex-differential manner. Katie L. Flanagan is Director of Infectious Diseases for north/north-west Tasmania, an adjunct professor at the University of Tasmania and RMIT University and an adjunct associate professor at Monash University. She is Honorary Secretary of the Australasian Society for Infectious Diseases (ASID), Chair of the ASID Vaccination Special Interest Group and a member of the Australian Technical Advisory Group on Immunisation. Her current research focuses on using systems vaccinology to study the sex-differential and non-targeted effects of vaccines. Sabra L. Klein is a professor of molecular microbiology and immunology at the Johns Hopkins Bloomberg School of Public Health, Baltimore, USA. She is an expert on sex and gender differences in immune responses and susceptibility to infection. She is the immediate past president of the Organization for the Study of Sex Differences, a principal investigator of the Johns Hopkins Specialized Center for Research Excellence in sex and age differences in immunity to influenza and a co-director of the Johns Hopkins Center for Women’s Health, Sex, and Gender Research. Tobias R. Kollmann is a paediatric infectious disease clinician and systems vaccinologist at Telethon Kids Institute and Perth Children’s Hospital in Perth, Australia. His expertise centres on newborn infectious diseases, immune ontogeny and early-life vaccine responses, using cutting-edge technology and analytics to extract the most information out of the typically small biological samples obtainable in early life. David J. Lynn is Director of the Computational and Systems Biology Program and an EMBL Australia group leader at the South Australian Health and Medical Research Institute. He is also a professor at the Flinders University College of Medicine and Public Health. He leads a research programme in systems immunology, investigating how pathogenic and commensal microorganisms modulate the immune system in different contexts, including vaccination. Frank Shann worked as a paediatrician in Papua New Guinea and then for 20 years was Director of Intensive Care at the Royal Children’s Hospital in Melbourne, Australia. He is a professorial fellow in the Department of Paediatrics, University of Melbourne, engaged in research on the non-specific effects of vaccines.
Collapse
Affiliation(s)
- Peter Aaby
- Bandim Health Project, Bissau, Guinea-Bissau.
| | - Christine Stabell Benn
- Bandim Health Project, OPEN, Department of Clinical Research, Odense University Hospital, Odense, Denmark. .,Danish Institute for Advanced Study, University of Southern Denmark, Odense, Denmark.
| | - Katie L Flanagan
- University of Tasmania, Hobart, TAS, Australia. .,RMIT University, Melbourne, VIC, Australia. .,Monash University, Melbourne, VIC, Australia.
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| | | | - David J Lynn
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia. .,Flinders University, Adelaide, SA, Australia.
| | - Frank Shann
- Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
122
|
Gebhard C, Regitz-Zagrosek V, Neuhauser HK, Morgan R, Klein SL. Impact of sex and gender on COVID-19 outcomes in Europe. Biol Sex Differ 2020; 11:29. [PMID: 32450906 PMCID: PMC7247289 DOI: 10.1186/s13293-020-00304-9] [Citation(s) in RCA: 726] [Impact Index Per Article: 145.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 04/24/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Emerging evidence from China suggests that coronavirus disease 2019 (COVID-19) is deadlier for infected men than women with a 2.8% fatality rate being reported in Chinese men versus 1.7% in women. Further, sex-disaggregated data for COVID-19 in several European countries show a similar number of cases between the sexes, but more severe outcomes in aged men. Case fatality is highest in men with pre-existing cardiovascular conditions. The mechanisms accounting for the reduced case fatality rate in women are currently unclear but may offer potential to develop novel risk stratification tools and therapeutic options for women and men. CONTENT The present review summarizes latest clinical and epidemiological evidence for gender and sex differences in COVID-19 from Europe and China. We discuss potential sex-specific mechanisms modulating the course of disease, such as hormone-regulated expression of genes encoding for the severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) entry receptors angiotensin converting enzyme (ACE) 2 receptor and TMPRSS2 as well as sex hormone-driven innate and adaptive immune responses and immunoaging. Finally, we elucidate the impact of gender-specific lifestyle, health behavior, psychological stress, and socioeconomic conditions on COVID-19 and discuss sex specific aspects of antiviral therapies. CONCLUSION The sex and gender disparities observed in COVID-19 vulnerability emphasize the need to better understand the impact of sex and gender on incidence and case fatality of the disease and to tailor treatment according to sex and gender. The ongoing and planned prophylactic and therapeutic treatment studies must include prospective sex- and gender-sensitive analyses.
Collapse
Affiliation(s)
- Catherine Gebhard
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland.
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland.
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria.
| | - Vera Regitz-Zagrosek
- University of Zurich, Zurich, Switzerland
- Charité, Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Hannelore K Neuhauser
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Robert Koch Institute, Berlin, Germany
| | - Rosemary Morgan
- Department of International Health, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
123
|
Abstract
Immunotherapies are often used for the treatment, remission, and possible cure of autoimmune diseases, infectious diseases, and cancers. Empirical evidence illustrates that females and males differ in outcomes following the use of biologics for the treatment of autoimmune diseases, e.g., rheumatoid arthritis (RA), infectious diseases, e.g., influenza, and solid tumor cancers. Females tend to experience more adverse reactions than males following the use of a class of biologics referred to as immunotherapies. For immunotherapies aimed at stimulating an immune response, e.g., influenza vaccines, females develop greater responses and may experience greater efficacy than males. In contrast, for immunotherapies that repress an immune response, e.g., tumor necrosis factor (TNF) inhibitors for RA or checkpoint inhibitors for melanoma, the efficacy is reportedly greater for males than females. Despite these differences, discrepancies in reporting differences between females and males exist, with females have been historically excluded from biomedical and clinical studies. There is a critical need for research that addresses the biological (i.e., sex) as well as sociocultural (i.e., gender) causes of male-female disparities in immunotherapy responses, toxicities, and outcomes. One-size-fits-all approaches to immunotherapies will not work, and sex/gender may contribute to variable treatment success, including adherence, in clinical settings.
Collapse
|
124
|
Bufan B, Arsenović-Ranin N, Petrović R, Živković I, Stoiljković V, Leposavić G. Strain specificities in influence of ageing on germinal centre reaction to inactivated influenza virus antigens in mice: Sex-based differences. Exp Gerontol 2020; 133:110857. [PMID: 32006634 DOI: 10.1016/j.exger.2020.110857] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 12/04/2019] [Accepted: 01/28/2020] [Indexed: 12/21/2022]
Abstract
Considering variability in vaccine responsiveness across human populations, in respect to magnitude and quality, and importance of vaccines in the elderly, the influence of recipient genetic background on the kinetics of age-related changes in the serum IgG antibody responses to seasonal trivalent inactivated split-virus influenza bulk (TIV) was studied in BALB/c and C57BL/6 mice showing quantitative and qualitative differences in this responses in young adult ages. With ageing the total serum IgG response to influenza viruses declined, in a strain-specific manner, so the strain disparity observed in young adult mice (the greater magnitude of IgG response in BALB/c mice) disappeared in aged mice. However, the sexual dimorphisms in this response (more prominent in females of both strains) remained in aged ones. The strain-specific differences in age-related decline in the magnitude of IgG response to TIV correlated with the number of germinal centre (GC) B splenocytes. The age-related decline in GC B cell number was consistent with the decrease in the proliferation of B cells and CD4+ cells in splenocyte cultures upon restimulation with TIV. Additionally, the age-related decrease in the magnitude of IgG response correlated with the increase in follicular T regulatory (fTreg)/follicular T helper (fTh) and fTreg/GC B splenocyte ratios (reflecting decrease in fTh and GC B numbers without changes in fTreg number), and the frequency of CD4+ splenocytes producing IL-21, a key factor in balancing the B cell and fTreg cell activity. With ageing the avidity of virus influenza-specific antibody increased in females of both strains. Moreover, ageing affected IgG2a/IgG1 and IgG2c/IgG1 ratios (reflecting Th1/Th2 balance) in male BALB/c mice and female C57BL/6 mice, respectively. Consequently, differently from young mice exhibiting the similar ratios in male and female mice, in aged female mice of both strains IgG2a(c)/IgG1 ratios were shifted towards a less effective IgG1 response (stimulated by IL-4 cytokines) compared with males. The age-related alterations in IgG subclass profiles in both strains correlated with those in IFN-γ/IL-4 production level ratio in splenocyte cultures restimulated with TIV. These findings stimulate further research to formulate sex-specific strategies to improve efficacy of influenza vaccine in the elderly.
Collapse
Affiliation(s)
- Biljana Bufan
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Nevena Arsenović-Ranin
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Raisa Petrović
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Irena Živković
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Vera Stoiljković
- Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Gordana Leposavić
- Department of Pathobiology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia.
| |
Collapse
|
125
|
Tadount F, Doyon-Plourde P, Rafferty E, MacDonald S, Sadarangani M, Quach C. Is there a difference in the immune response, efficacy, effectiveness and safety of seasonal influenza vaccine in males and females? - A systematic review. Vaccine 2019; 38:444-459. [PMID: 31711676 DOI: 10.1016/j.vaccine.2019.10.091] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/22/2019] [Accepted: 10/27/2019] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Seasonal influenza is an important cause of morbidity and mortality, despite being vaccine-preventable. Sex factors (genes and hormones) seem to impact individuals' susceptibility to infectious diseases and their response to vaccination. However, most vaccine studies do not explicitly assess sex differences in vaccine response, but rather adjust for sex. METHODS We conducted a systematic review to analyze immunogenicity, efficacy, effectiveness and/or safety of seasonal influenza vaccine data stratified by sex. We searched PubMed, EMBASE, CINAHL, Web of Science and clinicaltrials.gov for observational studies and phase III/IV trials from January 1990 to June 2018, published in English or French. Two reviewers independently screened all references, then proceeded to data extraction and quality assessment using the Cochrane tools (RoB and ROBINS-I) on included studies. RESULTS Of the 5,745 citations retrieved, 46 studies were included in the SR. Overall, 18 studies assessed immunogenicity, 1 estimated efficacy, 6 measured effectiveness and 25 evaluated safety of seasonal influenza vaccine in females and males (four studies reported on two sex-stratified outcomes concomitantly). CONCLUSION No clear conclusion could be drawn regarding the effect of sex on the immunogenicity and effectiveness of seasonal influenza vaccine, but higher rates of adverse events following immunization (AEFIs) were reported in females. The heterogeneity of data and studies' low quality prevented us from conducting a meta-analysis. There is a need to emphasize on the appropriate use of the terms sex and gender in biomedical research. Evidence of higher quality is needed to better understand sex differences in response to influenza vaccine.
Collapse
Affiliation(s)
- Fazia Tadount
- Department of Microbiology, Infectious Diseases, and Immunology, Faculty of Medicine, University of Montreal (QC) Canada; Research Institute - CHU Sainte Justine, Montreal (QC) Canada
| | - Pamela Doyon-Plourde
- Department of Microbiology, Infectious Diseases, and Immunology, Faculty of Medicine, University of Montreal (QC) Canada; Research Institute - CHU Sainte Justine, Montreal (QC) Canada
| | - Ellen Rafferty
- Faculty of Nursing, University of Alberta, Edmonton (AB) Canada
| | | | - Manish Sadarangani
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, Vancouver (BC) Canada; Division of Infectious Diseases, Department of Pediatrics, University of British Columbia, Vancouver (BC) Canada
| | - Caroline Quach
- Department of Microbiology, Infectious Diseases, and Immunology, Faculty of Medicine, University of Montreal (QC) Canada; Research Institute - CHU Sainte Justine, Montreal (QC) Canada; Department of Pediatric Laboratory Medicine, CHU Sainte-Justine, Montreal (QC) Canada; Infection Prevention & Control, CHU Sainte-Justine, Montreal (QC) Canada.
| |
Collapse
|
126
|
Dhakal S, Klein SL. Host Factors Impact Vaccine Efficacy: Implications for Seasonal and Universal Influenza Vaccine Programs. J Virol 2019; 93:e00797-19. [PMID: 31391269 PMCID: PMC6803252 DOI: 10.1128/jvi.00797-19] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Influenza is a global public health problem. Current seasonal influenza vaccines have highly variable efficacy, and thus attempts to develop broadly protective universal influenza vaccines with durable protection are under way. While much attention is given to the virus-related factors contributing to inconsistent vaccine responses, host-associated factors are often neglected. Growing evidences suggest that host factors including age, biological sex, pregnancy, and immune history play important roles as modifiers of influenza virus vaccine efficacy. We hypothesize that host genetics, the hormonal milieu, and gut microbiota contribute to host-related differences in influenza virus vaccine efficacy. This review highlights the current insights and future perspectives into host-specific factors that impact influenza vaccine-induced immunity and protection. Consideration of the host factors that affect influenza vaccine-induced immunity might improve influenza vaccines by providing empirical evidence for optimizing or even personalizing vaccine type, dose, and use of adjuvants for current seasonal and future universal influenza vaccines.
Collapse
Affiliation(s)
- Santosh Dhakal
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Sabra L Klein
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
127
|
Vom Steeg LG, Attreed SE, Zirkin B, Klein SL. Testosterone treatment of aged male mice improves some but not all aspects of age-associated increases in influenza severity. Cell Immunol 2019; 345:103988. [PMID: 31540670 PMCID: PMC6876866 DOI: 10.1016/j.cellimm.2019.103988] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 09/12/2019] [Accepted: 09/13/2019] [Indexed: 12/13/2022]
Abstract
The severity of influenza increases with age, with worse disease in aged males than females. Testosterone concentrations decline with age in males, which may impact influenza pathogenesis. Aged male mice were treated with testosterone or placebo and outcomes during influenza A virus (IAV) infection were compared with adult male mice. Aged males experienced greater morbidity and mortality than adult males, which was partially improved by testosterone treatment of aged males. Aged males cleared IAV from lungs slower than adult males, regardless of testosterone treatment. As compared with adult males, aged males experienced pulmonary, but not systemic, cytokine dysregulation, and delayed influx and contraction of IAV-specific CD8+ T cells in the lungs. Testosterone treatment in aged males partially restored pulmonary cytokine responses to levels consistent with adult males but did not alter the age-associated changes in IAV-specific CD8+ T cells. Testosterone only modestly improves outcomes of influenza in aged males.
Collapse
Affiliation(s)
- Landon G Vom Steeg
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sarah E Attreed
- Department of Environmental Health and Engineering, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Barry Zirkin
- Department of Biochemistry and Molecular Biology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Department of Biochemistry and Molecular Biology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
128
|
Host Factors Impact Vaccine Efficacy: Implications for Seasonal and Universal Influenza Vaccine Programs. J Virol 2019. [PMID: 31391269 DOI: 10.1128/jvi.00797‐19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Influenza is a global public health problem. Current seasonal influenza vaccines have highly variable efficacy, and thus attempts to develop broadly protective universal influenza vaccines with durable protection are under way. While much attention is given to the virus-related factors contributing to inconsistent vaccine responses, host-associated factors are often neglected. Growing evidences suggest that host factors including age, biological sex, pregnancy, and immune history play important roles as modifiers of influenza virus vaccine efficacy. We hypothesize that host genetics, the hormonal milieu, and gut microbiota contribute to host-related differences in influenza virus vaccine efficacy. This review highlights the current insights and future perspectives into host-specific factors that impact influenza vaccine-induced immunity and protection. Consideration of the host factors that affect influenza vaccine-induced immunity might improve influenza vaccines by providing empirical evidence for optimizing or even personalizing vaccine type, dose, and use of adjuvants for current seasonal and future universal influenza vaccines.
Collapse
|
129
|
Abstract
Introduction: High variance is associated with influenza vaccine effectiveness (VE). Accumulating evidence suggests that preexisting influenza-specific immunity results in the variance in VE and skews overall immune response to vaccination. Nevertheless, the investigation of preexisting immunity is highly limited due to the lack of proper methodology to explore the complex individual immune history.Areas covered: Retrospective observational studies have shown that the preexisting influenza specific immunity influences on VE. To simplify a discussion, we summarized important findings from the observational studies based on the transition of the individual immune history: the first exposure to influenza virus, the first vaccination, and repetitive exposure throughout life. We also discussed the prospectus of pre-immunized animal models to investigate the interaction between preexisting immunity and vaccine efficacy.Expert opinion: A better understanding in the underlying mechanisms on preexisting immunity is critical to improve VE and to help develop novel vaccine strategies. Using animals pre-immunized with historical influenza strains is a promising approach to verify the underlying immunologic mechanism of interaction between preexisting immunity and vaccine antigen. Also, pre-immunized animal models will be better able to evaluate the efficacy of novel vaccine strategies than naïve animals.
Collapse
Affiliation(s)
- Hyesun Jang
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | - Ted M Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA.,Department of Infectious Diseases, University of Georgia, Athens, GA, USA
| |
Collapse
|